CA2877473A1 - Imidazo bicyclic imminium compounds as antitumor agents - Google Patents

Imidazo bicyclic imminium compounds as antitumor agents Download PDF

Info

Publication number
CA2877473A1
CA2877473A1 CA2877473A CA2877473A CA2877473A1 CA 2877473 A1 CA2877473 A1 CA 2877473A1 CA 2877473 A CA2877473 A CA 2877473A CA 2877473 A CA2877473 A CA 2877473A CA 2877473 A1 CA2877473 A1 CA 2877473A1
Authority
CA
Canada
Prior art keywords
optionally substituted
alkyl
chosen
phenyl
fused
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2877473A
Other languages
French (fr)
Inventor
James Chen
Tomoyo Sakata KATO
Alison Evelynn ONDRUS
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Leland Stanford Junior University
Original Assignee
Leland Stanford Junior University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Leland Stanford Junior University filed Critical Leland Stanford Junior University
Publication of CA2877473A1 publication Critical patent/CA2877473A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/429Thiazoles condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53831,4-Oxazines, e.g. morpholine ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Imidazobicycles having a quaternary nitrogen, i.e. imminium salts, are disclosed. Examples include derivatives of 6,7,8,9-tetrahydro-5H-imidazo[1,2-a]azepinium and 2,3-dihydroimidazo[2,1-b]thiazolium salts. The compounds are inhibitors of the Hedgehog pathway and therefore useful as antitumor agents and as probes of the function of Hedgehog-dependent systems.

Description

IMIDAZO BICYCLIC IMMINIUM COMPOUNDS AS
ANTITUMOR AGENTS
FEDERALLY SPONSORED RESEARCH
[0001] This invention was made with Government support under contract CA136574 awarded by the National Institutes of Health. The Government has certain rights in this invention.
CROSS-REFERENCE TO RELATED APPLICATIONS
[0002] This application claims priority from US provisional application 61/662,977, filed June 22, 2012, the entire contents of which are incorporated herein by reference.
FIELD OF THE INVENTION
[0003] The invention relates to imidazobicycles having a quaternary nitrogen that are inhibitors of the Hedgehog pathway and therefore useful as antitumor agents and as probes of the function of Hedgehog-dependent systems.
BACKGROUND OF THE INVENTION
[0004] The Hedgehog (Hh) pathway plays a critical role in the patterning, homeostasis, and oncogenic transformation of multiple tissues. For example, Hh signaling regulates cerebellar patterning and growth, and Hh pathway activation is a leading cause of medulloblastoma, the most common pediatric brain cancer. Genetic screens have revealed a number of Hh pathway regulators, including canonical signaling proteins that are conserved across metazoans and vertebrate-specific modulators. These studies have provided a general framework for vertebrate Hh signal transduction, which in mammals is initiated by the binding of secreted polypeptides (Sonic, Shh; Indian, Ihh; or Desert, Dhh) to the 12-transmembrane receptor Patchedl (Ptchl) and the subsequent activation of Smoothened (Smo), a G protein-coupled receptor-like protein. Smo then acts through unknown mechanisms to control the functions of G1i2 and G1i3, zinc finger transcription factors that exist as a balance between N-terminal repressors (G1i2/3R), full-length proteins (G1i2/3FL), or phosphorylated forms of G1i2/3FL that are transcriptionally active (G1i2/3A). This process is mediated at least in part by Suppressor of Fused (Sufu), a direct negative regulator of Gli function. The primary cilium serves as a Hh pathway nexus; Ptchl, Smo, Sufu, G1i2, and G1i3 traffic through the cilium in a pathway activity-dependent manner, and many of their interactions appear to occur within or depend upon this microtubule-scaffolded structure. Upon Hh pathway activation, G1i2A and to a lesser extent G1i3A then drive the transcription of Hh target genes, including Ptchl and the constitutively active factor Glil.
[0005] The therapeutic utility of Hh pathway inhibitors has been confirmed by the recent approval of vismodegib (ERIVEDGETM) by the U.S. Food and Drug Administration.
It is currently indicated for patients with basal cell carcinoma. Vismodegib is also undergoing clinical trials for metastatic colorectal cancer, small-cell lung cancer, advanced stomach cancer, pancreatic cancer, chondrosarcoma and medulloblastoma.
Vismodegib acts as an antagonist of the Smoothened receptor (SMO). SMO inhibition causes the transcription factors GLI 1 and GLI2 to remain inactive, which prevents the expression of tumor-mediating genes within the Hedgehog pathway. This pathway is pathogenetically relevant in more than 90% of basal cell carcinomas.
[0006] Current SMO-targeting therapies, despite their promise, have some potential drawbacks. Hh signaling is an important regulator of bone growth in juvenile mice, and Smo inhibitors cause permanent dwarfism in animal models. While such pharmacological side effects may be outweighed by the pernicious nature of certain Hh pathway-induced cancers, the side effects may warrant contraindication in other cases. New Hh pathway inhibitors, including those that act through different mechanisms, are needed to address these challenges.
[0007] In addition to their potential utility in treating tumors, small-molecule inhibitors of the Hh pathway can be valuable tools for studying Hh signaling mechanisms.
SUMMARY OF THE INVENTION
[0008] In one aspect, the invention relates to a method of inhibiting Hedgehog pathway activation comprising bringing a cell that is capable of Hedgehog expression into contact with a compound of formula I:

R2,.,.,.,( X-----' zN:L--....zssyNrD
W A
I
wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2 is chosen from H, halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, optionally substituted aryl and optionally substituted heteroaryl;
R3 is chosen from H, alkyl, optionally substituted aryl and optionally substituted heteroaryl;
A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (Ci-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (Ci-C4)alkyl residues;
and X is any counterion. In these compounds, one of R2 and R3 must be optionally substituted aryl or optionally substituted heteroaryl.
[0009] In another aspect, the invention relates to the use of a compound of formula I for inhibiting the growth of a solid tumor.
[0010] In another aspect, the invention relates to method of probing Hedgehog function in vitro comprising bringing a cell that is capable of Hedgehog expression into contact with a compound of formula I.
[0011] In another aspect, the invention relates to a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of formula II:
R3a R2z..........N.,...õ,...,( X-W Al ..--------z4----) II
wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2a is chosen from H and (Ci-C4)alkyl;
R3' is chosen from optionally substituted aryl and optionally substituted heteroaryl;
A1 is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (Ci-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (Ci-C4)alkyl residues;
and X is any counterion.
[0012] In another aspect, the invention relates to compounds of formula III:

/ \ R
--------X---------µ

N''.......0A

III
wherein A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (Ci-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (Ci-C4)alkyl residues;
R6 is one or two substituents chosen independently from the group consisting of halogen, halo (C i-C6)alkyl, (Ci-C6)alkyl, (Ci-C6)acyl, (Ci-C6)alkoxy(Ci-C6)alkyl, hydroxy(Ci-C6)alkyl, phenyl, benzenesulfonyl, hydroxy, (Ci-C6)alkoxy, halo(Ci-C6)alkoxy, oxaalkyl, carboxy, (Ci-C6)alkoxycarbonyl , (Ci-C6)alkoxycarbonylamino, carboxamido, cyano, acetoxy, nitro, amino, (Ci-C6)alkylamino, (Ci-C6)dialkylamino, mercapto, (C1-C6)alkylthio, (Ci-C6)alkylsulfoxide, (C1-C6)alkylsulfonyl, benzyl, heterocyclyl, phenoxy, heteroaryloxy and benzyloxy, and X is any counterion.

DETAILED DESCRIPTION OF THE INVENTION
[0013] The antitumor compounds described herein inhibit Hedgehog signaling and thereby inhibit tumor growth. The compounds that are useful in the methods described herein fall into a genus of formula I:

R2.,,..,.,( X-W A
(I) In these compounds, R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl. In some embodiments, R1 is phenyl or optionally substituted phenyl. For example, R1 may be phenyl or phenyl substituted with a substituent chosen from halogen, (Ci-C4)alkyl, halo (Ci-C4)alkyl, (Ci-C4)alkoxy, halo (Ci-C4)alkoxy, (Ci-C2)alkylenedioxy and phenyl.
[0014] R2 is chosen from H, halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, optionally substituted aryl and optionally substituted heteroaryl. When R2 is H, halogen, (Ci-C4)alkyl or halo(Ci-C4)alkyl, then R3 must be optionally substituted aryl or optionally substituted heteroaryl.
[0015] R3 may be chosen from H, alkyl, optionally substituted aryl and optionally substituted heteroaryl. In some embodiments R2 is H or methyl and R3 is phenyl or phenyl substituted with a substituent chosen from halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, (Ci-C2)alkylenedioxy, and phenyl. In some embodiments, R3 is phenyl substituted with a substituent chosen from halogen, halo(Ci-C6)alkyl, (Ci-C6)alkyl, (Ci-C6)acyl, (Ci-C6)alkoxy(Ci-C6)alkyl, hydroxy(Ci-C6)alkyl, phenyl, benzenesulfonyl, hydroxy, (Ci-C6)alkoxy, halo(Ci-C6)alkoxy, oxaalkyl, carboxy, (Ci-C6)alkoxycarbonyl , (C1-C6)alkoxycarbonylamino, carboxamido, cyano, acetoxy, nitro, amino, (Ci-C6)alkylamino, (Ci-C6)dialkylamino, mercapto, (Ci-C6)alkylthio, (Ci-C6)alkylsulfoxide, (C1-C6)alkylsulfonyl, benzyl, heterocyclyl, phenoxy, heteroaryloxy and benzyloxy. In other embodiments, R3 is H or methyl and R2 is phenyl or phenyl substituted with a substituent chosen from halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, (Ci-C2)alkylenedioxy, and phenyl.
[0016] Ring A may be a fused, saturated ring of 5 to 7 members. The ring may contain other heteroatoms in addition the nitrogen at the point of fusion, and it may be optionally substituted with one or two (Ci-C4)alkyl residues. In addition, ring A may be a fused bicycle, at least one ring of the fused bicycle being non-aromatic. The bicycle may be optionally substituted with one or two (Ci-C4)alkyl residues in either of its rings and may contain additional heteroatoms in either of its rings. In some embodiments, ring A is chosen from pyrrolidine, piperidine, azepine, thiazolidine, oxazolidine, imidazolidine, thiazine, oxazine, piperazine, oxazepine, thiazepine, diazepine and tetrahydroquinoline. In some embodiments, ring A is chosen from pyrrolidine, piperidine, azepine, thiazolidine, thiazine, morpholine and tetrahydroquinoline. For example, when ring A is azepine, the compounds of formula I are derivatives of 6,7,8,9-tetrahydro-5H-imidazo[1,2-a]azepinium:

R2N...........K X-N
zNr----,............0 wherein R4 is one or two (Ci-C4)alkyl residues. Similarly, when ring A is thiazole, the compounds of formula I are derivatives of 2,3-dihydroimidazo[2,1-b]thiazolium:

R2N............. X-------N
Nt.--=-........< ------1 V _R4 W
..---j S

wherein R4 is one or two (Ci-C4)alkyl residues.
[0017] The compounds of the invention are imminium derivatives, i.e. cationic species.
Therefore they will always be presented as salts, and the term "pharmaceutically acceptable salt" refers to salts whose counter ion (anion) derives from pharmaceutically acceptable non-toxic acids including inorganic acids, organic acids and water (which formally furnishes the hydroxide anion). Suitable pharmaceutically acceptable anions for the compounds of the present invention include hydroxide, acetate, benzenesulfonate (besylate), benzoate, bicarbonate, bisulfate, carbonate, camphorsulfonate, citrate, ethanesulfonate, fumarate, gluconate, glutamate, glycolate, bromide, chloride, isethionate, lactate, maleate, malate, mandelate, methanesulfonate, mucate, nitrate, pamoate, pantothenate, phosphate, succinate, sulfate, tartrate, trifluoroacetate, p-toluenesulfonate, acetamidobenzoate, adipate, alginate, aminosalicylate, anhydromethylenecitrate, ascorbate, aspartate, calcium edetate, camphorate, camsylate, caprate, caproate, caprylate, cinnamate, cyclamate, dichloroacetate, edetate (EDTA), edisylate, embonate, estolate, esylate, fluoride, formate, gentisate, gluceptate, glucuronate, glycerophosphate, glycolate, glycollylarsanilate, hexylresorcinate, hippurate, hydroxynaphthoate, iodide, lactobionate, malonate, mesylate, napadisylate, napsylate, nicotinate, oleate, orotate, oxalate, oxoglutarate, palmitate, pectinate, pectinate polymer, phenylethylbarbiturate, picrate, pidolate, propionate, rhodanide, salicylate, sebacate, stearate, tannate, theoclate, tosylate and the like. The desired salt may be obtained by ion exchange of whatever counter ion is obtained in the synthesis of the imminium compound. These methods are well known to persons of skill. Although pharmaceutically acceptable counter ions will be preferred for preparing pharmaceutical formulations and for use in therapeutic methods, other anions are quite acceptable as synthetic intermediates.
Thus the anion may be pharmaceutically undesirable when such salts are chemical intermediates. For the purpose of therapeutic methods and pharmaceutical compositions, it is desirable that the counterion designated X- be a pharmaceutically acceptable anion.
[0018] All of the compounds falling within the foregoing parent genus and its subgenera are useful as modulators of the Hedgehog pathway and are useful for treating the cancers described below. A search of the literature indicates that certain compounds useful in the methods described herein have been disclosed for the treatment of inflammatory or obstructive airways diseases. [See US published application 2008/0207718, which discloses compounds in which ring A is pyrrolidine.] For that reason, the genus of pharmaceutical compositions is smaller than the chemical genus of compounds useful in the methods. In a composition aspect, the invention relates to a pharmaceutically acceptable carrier and a compound of formula II:
R3a R2............s....( X-..---"--..
W
z(0 Al II
[0019] In some embodiments, R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl. In some embodiments, R1 is phenyl or phenyl substituted with a substituent chosen from halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, (Ci-C2)alkylenedioxy and phenyl.
[0020] In some embodiments, R2a is H; in others R2a is (Ci-C4)alkyl, such as methyl or ethyl.
[0021] In some embodiments, R3' is optionally substituted aryl or optionally substituted heteroaryl. In some embodiments, R3a is phenyl or phenyl substituted with a substituent chosen from halogen, (Ci-C4)alkyl, halo(Ci-C4)alkyl, (Ci-C4)alkoxy, halo(Ci-C4)alkoxy, (Ci-C2)alkylenedioxy, and phenyl.
[0022] Al is a fused, saturated ring of 6 or 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, and the saturated ring may be substituted with one or two (Ci-C4)alkyl residues. In some embodiments, ring Al may be piperidine, azepine, thiazine, oxazine, piperazine, oxazepine, thiazepine or diazepine. In some embodiments, ring A1 is azepine and a subgenus of II has the formula Ha:
R3a R2 ...õ.....K X--------N
zN*---,..........0 ha.
[0023] In other embodiments, ring A1 is piperidine and a subgenus of II has the formula lib:
R3a R2..............õ( X-..-/---.
N
V

Ith.
[0024] In other embodiments, ring A1 is tetrahydroquinoline and a subgenus of II has the formula He:
R3a R2.T.N______K
------.
N

V
W

He
[0025] As noted above, all of the compounds falling within the parent genus I
and its subgenera are useful as modulators of the Hedgehog pathway and are useful for treating the cancers described below. A search of the literature further indicates that certain compounds useful in the methods described herein have been disclosed without utility, generally from synthetic libraries, in Chemical Abstracts. Excluding such compounds, a subgenus, herein designated III, appears both novel and unexpectedly active.
[0026] Throughout this specification the terms and substituents retain their definitions.
[0027] Alkyl is intended to include linear, branched and cyclic hydrocarbon structures and combinations thereof A combination would be, for example, cyclopropylmethyl. Any hydrocarbon in which all carbons are essentially sp3 hybridized and no carbons are sp2 or sp hybridized is considered alkyl. To be perfectly clear, when a substituent is (Ci-C6)alkyl, it is meant that it can be a straight chain (for instance, methyl or ethyl), a branched chain (e.g., t-butyl), a cycloalkyl (for instance, cyclopropyl or cyclobutyl), or a combination (e.g., methylcyclopropyl). If a substituent is described more specifically, however, it takes on that definition; for instance, recitation of "cycloalkyl" refers only to a cyclic alkyl and not a linear or combination alkyl. Lower alkyl refers to alkyl groups of from 1 to 6 carbon atoms.
Examples of lower alkyl groups include methyl, ethyl, propyl, isopropyl, cyclopropyl, butyl, s-and t-butyl, cyclobutyl and the like. Cycloalkyl is a subset of alkyl and includes cyclic hydrocarbon groups of from 3 to 8 carbon atoms. Examples of cycloalkyl groups include c-propyl, c-butyl, c-pentyl, norbornyl, adamantyl and the like.
[0028] Alkoxy or alkoxyl refers to alkyl groups of from 1 to 8 carbon atoms of a straight, branched, or cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. Lower-alkoxy refers to groups containing one to four carbons.
[0029] Aryl and heteroaryl mean a 5- or 6-membered aromatic or heteroaromatic ring containing 0-3 heteroatoms selected from 0, N, or S; a bicyclic 9- or 10-membered aromatic or heteroaromatic ring system containing 0-3 heteroatoms selected from 0, N, or S; or a tricyclic 13- or 14-membered aromatic or heteroaromatic ring system containing 0-3 heteroatoms selected from 0, N, or S. The aromatic 6- to 14-membered carbocyclic rings include, e.g., benzene, naphthalene, indane, tetralin, and fluorene and the 5-to 10-membered aromatic heterocyclic rings include, e.g., imidazole, pyridine, indole, thiophene, benzopyranone, thiazole, furan, benzimidazole, quinoline, isoquinoline, quinoxaline, pyrimidine, pyrazine, tetrazole and pyrazole. As used herein aryl and heteroaryl refer to residues in which one or more rings are aromatic, but not all need be.
[0030] Arylalkyl means an aryl ring attached to an alkyl residue in which the point of attachment to the parent structure is through the alkyl. Examples are benzyl, phenethyl and the like. Heteroarylalkyl means an a heteroaryl ring attached through an alkyl residue to the parent structure. Examples include, e.g., pyridinylmethyl, pyrimidinylethyl and the like.
[0031] C1 to C10 hydrocarbon means a linear, branched, or cyclic residue comprised of hydrogen and carbon as the only elemental constituents and includes alkyl, cycloalkyl, polycycloalkyl, alkenyl, alkynyl, aryl and combinations thereof Examples include benzyl, phenethyl, cyclohexylmethyl, cyclopropylmethyl, cyclobutylmethyl, allyl and camphoryl.
[0032] Acyl refers to formyl and to groups of 1, 2, 3, 4, 5, 6, 7 and 8 carbon atoms of a straight, branched, cyclic configuration, saturated, unsaturated and aromatic and combinations thereof, attached to the parent structure through a carbonyl functionality.
One or more carbons in the acyl residue may be replaced by nitrogen, oxygen or sulfur as long as the point of attachment to the parent remains at the carbonyl.
Examples include acetyl, benzoyl, propionyl, isobutyryl, t-butoxycarbonyl, benzyloxycarbonyl and the like.
Lower-acyl refers to groups containing one to four carbons.
[0033] Unless otherwise specified, the term "carbocycle" is intended to include ring systems in which the ring atoms are all carbon but of any oxidation state.
Thus (C3-C1o) carbocycle refers to both non-aromatic and aromatic systems, including such systems as cyclopropane, cyclobutane, cyclopentane, cyclohexane, benzene, cyclohexene and cyclohexadiene; (C8-C12) carbopolycycle refers to such systems as norbornane, decalin, indane, adamantane and naphthalene. Carbocycle, if not otherwise limited, refers to monocycles, bicycles and polycycles.
[0034] Heterocycle means a cycloalkyl or aryl residue in which one to three of the carbons is replaced by a heteroatom such as oxygen, nitrogen or sulfur.
Heteroaryls form a subset of heterocycles. Examples of heterocycles include pyrrolidine, pyrazole, pyrrole, imidazole, indole, quinoline, isoquinoline, tetrahydroisoquinoline, benzofuran, benzodioxan, benzodioxole (commonly referred to as methylenedioxyphenyl, when occurring as a substituent), tetrazole, morpholine, thiazole, pyridine, pyridazine, pyrimidine, pyrazine, thiophene, furan, oxazole, oxazoline, isoxazole, dioxane, tetrahydrofuran and the like.
[0035] As used herein, the term "optionally substituted" may be used interchangeably with "unsubstituted or substituted". The term "substituted" refers to the replacement of one or more hydrogen atoms in a specified group with a specified substituent. For example, substituted alkyl, aryl, cycloalkyl, heterocyclyl etc. refer to alkyl, aryl, cycloalkyl, or heterocyclyl wherein one or more H atoms in each residue are replaced with halogen, haloalkyl, alkyl, acyl, alkoxyalkyl, hydroxyloweralkyl, carbonyl, phenyl, heteroaryl, benzenesulfonyl, hydroxy, loweralkoxy, haloalkoxy, oxaalkyl, carboxy, alkoxycarbonyl [-C(=0)0-alkyl], alkoxycarbonylamino [ HNC(=0)0-alkyl], carboxamido [-C(=0)NH2], alkylaminocarbonyl [-C(=0)NH-alkyl], cyano, acetoxy, nitro, amino, alkylamino, dialkylamino, mercapto, alkylthio, alkylsulfinyl, alkylsulfonyl, benzyl, phenoxy, and benzyloxy. Although in most cases of "optionally substituted" residues, 1, 2 or 3 hydrogen atoms are replaced with a specified substituent, in the case of fluoroalkyl residues, more than three hydrogen atoms can be replaced by fluorine; indeed, all available hydrogen atoms could be replaced by fluorine, e.g. perfluoropropyl. In most cases, Ci to C6 alkyl residues or C1 to C4 alkyl residues are preferred as sub stituents.
[0036] The compounds described herein may contain one or more asymmetric centers in ring A and/or in the Rx groups, and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)-. The present invention is meant to include all such possible isomers, as well as their racemic and optically pure forms. Optically active (R)- and (S)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques. When the compounds described herein contain olefinic double bonds or other centers of geometric asymmetry, and unless specified otherwise, it is intended that the compounds include both E and Z geometric isomers.
[0037] Likewise, all tautomeric forms and resonance structures are also intended to be included. For example, the structural representation of subgenus Ha can be presented either as the exocyclic imminium ion or the endocyclic imminium ion:

R3a R3a R2.....,...K X- R2eK X-------- ------N
R1 ......ON+
,N

Or =
The two are equivalent.
[0038] As used herein, and as would be understood by the person of skill in the art, the recitation of "a compound" - unless expressly further limited - is intended to include salts of that compound. In a particular embodiment, the term "compound of formula I"
refers to the pharmaceutically acceptable salt.
[0039] It will be recognized that the compounds of this invention can exist in radiolabeled form, i.e., the compounds may contain one or more atoms containing an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
Alternatively, a plurality of molecules of a single structure may include at least one atom that occurs in an isotopic ratio that is different from the isotopic ratio found in nature.
Radioisotopes of hydrogen, carbon, phosphorous, fluorine, chlorine and iodine include, for 2H 3H 11C 13C 14C 15N 35s 18F 3605 12315 12515 1311 and 133 example,I . Compounds that contain those radioisotopes and/or other radioisotopes of other atoms are within the scope of this invention. Compounds containing 3H, 14C and iodine radioisotopes are particularly preferred for their ease in preparation and detectability. Compounds that contain isotopes iic5 13N5 150 and 18F are well suited for positron emission tomography.
Radiolabeled compounds of formulae I and II of this invention can generally be prepared by methods well known to those skilled in the art. Conveniently, such radiolabeled compounds can be prepared by carrying out the procedures disclosed in the Examples and Schemes by substituting a readily available radiolabeled reagent for a non-radiolabeled reagent.
[0040] Although this invention is susceptible to embodiment in many different forms, preferred embodiments of the invention are shown. It should be understood, however, that the present disclosure is to be considered as an exemplification of the principles of this invention and is not intended to limit the invention to the embodiments illustrated.
[0041] When the compounds of formula I, II or III are to be employed as antitumor agents in vivo, they may be administered as the raw chemical, but it is preferable to present them as a pharmaceutical composition. According to a further aspect, the present invention provides a pharmaceutical composition comprising a compound of formula II or a pharmaceutically acceptable salt or solvate thereof, together with one or more pharmaceutically carriers thereof and optionally one or more other therapeutic ingredients.
The carrier(s) must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof The compositions may be formulated for oral, topical or parenteral administration.
For example, they may be given intravenously, intraarterially, subcutaneously, and directly into the CNS
¨ either intrathecally or intracerebroventricularly.
[0042] Formulations include those suitable for oral, parenteral (including subcutaneous, intradermal, intramuscular, intravenous and intraarticular), rectal and topical (including dermal, buccal, sublingual and intraocular) administration. The compounds are preferably administered orally or by injection (intravenous or subcutaneous). The precise amount of compound administered to a patient will be the responsibility of the attendant physician.
However, the dose employed will depend on a number of factors, including the age and sex of the patient, the precise disorder being treated, and its severity. Also, the route of administration may vary depending on the condition and its severity. The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product into the desired formulation.
[0043] Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil-in-water liquid emulsion or a water-in-oil liquid emulsion. The active ingredient may also be presented as a bolus, electuary or paste.
[0044] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricating, surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide sustained, delayed or controlled release of the active ingredient therein.
[0045] Formulations for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient.
Formulations for parenteral administration also include aqueous and non-aqueous sterile suspensions, which may include suspending agents and thickening agents. The formulations may be presented in unit-dose of multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of a sterile liquid carrier, for example saline, phosphate-buffered saline (PBS) or the like, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
[0046] Formulations for rectal administration may be presented as a suppository with the usual carriers such as cocoa butter or polyethylene glycol.
[0047] Formulations for topical administration in the mouth, for example buccally or sublingually, include lozenges comprising the active ingredient in a flavoured basis such as sucrose and acacia or tragacanth, and pastilles comprising the active ingredient in a basis such as gelatin and glycerin or sucrose and acacia.
[0048] Preferred unit dosage formulations are those containing an effective dose, or an appropriate fraction thereof, of the active ingredient.
[0049] It should be understood that in addition to the ingredients particularly mentioned above, the formulations of this invention may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.
[0050] As used herein, the terms "treatment" or "treating," or "palliating" or "ameliorating" refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological systems associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. The compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological systems of a disease, even though a diagnosis of this disease may not have been made.
[0051] The compounds employed in the methods described herein may be purchased from the following vendors: Life Chemicals, MolPort, Princeton Biomolecular Research Inc., and Sigma-Aldrich. Alternatively, they may be synthesized by methods well-known in the art.
For example, a series of papers from the laboratory of A. M. Demchenko describes the syntheses of many imidazobicycles having a quaternary nitrogen [e.g. (1) Russian Journal of General Chemistry (Translationof Zhurnal Obshchei Khimii) (2001), 71, 1759-1763; (2) Chemistry of Heterocyclic Compounds (Translation of Khimiya Geterotsiklicheskikh Soedinenii) (2003), 39,1084-1089; (3) Chemistry of Heterocyclic Compounds (2001), 37, 1054-1056; (4) Dopovidi Natsional'noi Akademii Nauk Ukraini (2000), 144-147;
(5) Pharmaceutical Chemistry Journal (Translation of Khimiko-Farmatsevticheskii Zhurnal) (1999), 33, 421-423; (6) Russian Journal of General Chemistry (1997), 67, 1775-1781; (7) Chemistry of Heterocyclic Compounds (1997), 33, 724-727; (8) Ukrainskii Khimicheskii Zhurnal (Russian Edition) (1996), 62, 42-47; and (9) Zhurnal Prikladnoi Khimii (Sankt-Peterburg) (1996), 69, 1501-1504]. The contents of these articles is incorporated herein by reference. In accordance with the teachings of Demchenko, 2-arylazacycle amidines are condensed with phenacyl halides to produce compounds of the genus I:

W A
N( Br 0 zN.,,...s.,(1D _)._ ,N-,....--;,,,...0 Br-For example:
Br Br .

Br 0 N...., Br-Et0 DO

In like manner the other compounds in the Table 1 below may be synthesized.
[0052] Preparation of compounds A-19 and A-18 1. 85 C, 6 d, 20%
2.
Me0 N OMe Me0 io NH2 Br fh, NO

THF, 78 C, 16 h; HPLC, 89% A-19
[0053] A mixture of o-anisidine (1.57 mL, 13.9 mmol, 1.00 equiv) and 7-methoxy-3,4,5,6-tetrahydro-2H-azepine (2.00 mL, 13.9 mmol, 1 equiv) was heated to 85 C and stirred under a nitrogen atmosphere for 6 days. The resulting tan precipitate was collected by vacuum filtration and washed with 3 x 3 mL Et20 to yield N-(2-methoxypheny1)-3,4,5,6-tetrahydro-2H-azepin-7-amine as a pale tan solid (608 mg, 20%).
[0054] 1-([1,1'-bipheny1]-4-y1)-2-bromoethan-1-one (126 mg, 0.458 mmol, 1.00 equiv) was added to a pressure vessel charged with a suspension of N-(2-methoxypheny1)-3,4,5,6-tetrahydro-2H-azepin-7-amine (100 mg, 0.458 mmol, 1 equiv) in THF (0.916 mL) and the vessel was sealed and placed on an oil bath at 78 C. The reaction mixture became a clear yellow solution within 15 min. The vessel was maintained at 78 C for 16 h, then allowed to cool to room temperature, and the solution was concentrated under reduced pressure to yield a pale brown oily residue.
[0055] The residue was dissolved in acetic anhydride (1.35 mL) in a pressure vessel, and the vessel was sealed and maintained on an oil bath at 120 C for 5 h. The vessel was then allowed to cool to room temperature, the deep brown solution was concentrated under reduced pressure, and the residue was purified by flash column chromatography on silica gel (eluent: gradient, 2.5 5%
Me0H in CH2C12, diam. 1.5 cm, ht. 6 cm) to yield A-18 (X
= Br) as a brown oil. Further purification by HPLC (Agilent Microsorb 300-5 Dynamax 250 x 21.4mm, gradient: 20 75% MeCN (0.1% TFA) in H20 (0.1% TFA), 35 min, 5 12.5 mL/min, tR = 12.5 min) provided A-19 (X = 02CCF3, 207 mg, 89%) as a pale tan oil.

1H NMR (400 MHz, CDC13, 25 C) 7.75 (d, J= 8.0 Hz, 2H, NCHCCCH), 7.45-7.63 (m, 8H), 7.39 (tt, J= 7.4, 1.2 Hz, 1H, (CH)2CH(CH)2), 7.11-7.16 (m, 2H, MeOCCH, MeOCCCHCH), 7.11 (s, 1H, NCHCN), 4.22-4.40 (m, 2H, CNCH2), 3.88 (s, 3H, OCH3), 2.90-3.04 (m, 2H, CH2CN), 1.64-2.10 (m, 6H, CH2(CH2)3CH2)=
13C NMR (100 MHz, CDC13, 25 C) 153.6, 150.9, 143.7, 139.8, 134.9, 133.2, 130.7, 130.7, 129.3, 129.3, 128.4, 128.4, 128.3, 128.3, 127.4, 127.4, 123.8, 122.7, 122.0, 119.9, 112.7,
56.3, 48.1, 29.7, 27.1, 25.5, 23.7.
[0056] A-18 was prepared according to the procedure described for A-19 ( X =
02C CF3 : 214 mg, yield: 98% from N-(2-methoxypheny1)-3,4,5,6-tetrahydro-2H-azepin-7-amine).
1H NMR (400 MHz, CDC13, 25 C) 7.55 (td, J= 7.9, 1.5 Hz, 1H, MeOCCCH), 7.45 (dd, J= 8.2, 1.5 Hz, 1H, MeOCCHCH), 7.34 (d, J= 8.6 Hz, 2H, NCHCCCH), 7.13 (d, J= 8.2 Hz, 1H, CH3OCCH), 7.10-7.14 (m, 1H, NCHCH), 7.00 (d, J= 8.6 Hz, 2H, CH3CH2OCCH), 7.01 (s, 1H, NCHCN), 4.24-4.29 (m, 2H, NCH2), 4.06 (q, J= 7.0 Hz, 2H, OCH2CH3), 3.86 (s, 3H, OCH3), 2.87-3.02 (m, 2H, CH2CN), 1.62-2.09 (m, 6H, CH2(CH2)3CH2), 1.41 (t, J= 7.0 Hz, 3H, OCH2CH3).
13C NMR (100 MHz, CDC13, 25 C) 160.8, 153.3, 150.1, 134.9, 132.9, 131.5, 131.5, 128.1, 122.5, 121.6, 119.2, 116.3, 115.3, 115.3, 112.4, 63.8, 56.0, 47.6, 29.4, 26.8, 25.3, 23.5, 14.6.
[0057] Assays and Test results
[0058] Sufu-KO-LIGHT cells were derived from Sufu knockout mouse embryonic fibroblasts according to the method of Chen et al. [Genes Dev.
2009;23(16):1910-28.] The cells were co-transfected with the zeocin resistance vector (pVgRXR, Invitrogen) and a firefly luciferase reporter driven by eight Gli binding sites and a y-crystallin basal promoter (8XG1iBS-FL) [see Sasaki et al. Development. 1997;124(7):1313-22.2]. Selection with 400 g/mL zeocin (Invitrogen, R250-01) and cell cloning were then conducted to generate the Sufu-KO-LIGHT line, which demonstrates constitutive Hh pathway activation that is sensitive to HPI-1 treatment. Sufu-KO-LIGHT cells were cultured in DMEM
(Invitrogen, 11965) containing 10% (v/v) fetal bovine serum (Invitrogen, No. 26140), 150 g/mL zeocin (Invitrogen, R250-01), 1 mM sodium pyruvate (Invitrogen, 11360) and 1X
PenStrep (Gibco, 15140).
[0059] Shh-LIGHT2 cells [see Taipale et al. Nature. 2000;406(6799):1005-9], an NIH-3T3-based cell line containing a stably integrated Gli-responsive firefly luciferase reporter (8XG1iBS-FL) and constitutive Renilla luciferase reporter (pRLTK, Promega), were cultured in DMEM (Invitrogen, 11965) containing 10% (v/v) calf serum [the American Type Culture Collection (ATCC), 30-2030], 150 g/mL zeocin (Invitrogen, R250-01), 400 g/mL G418 (Invitrogen, 11811), 1 mM sodium pyruvate (Invitrogen, 11360) and 1X

PenStrep.
[0060] C3H10T(1/2) cells were obtained from ATCC and cultured in DMEM
(Invitrogen, 11965) containing 10% (v/v) fetal bovine serum (Invitrogen, No. 26140), and 1X
PenStrep (Gibco, 15140).
[0061] NIH-3T3 cells were obtained from ATCC and cultured in DMEM (Invitrogen, 11965) containing 10% (v/v) calf serum (ATCC, 30-2030), 1 mM sodium pyruvate (Invitrogen, 11360) and lx PenStrep (Gibco, 15140).
[0062] Shh-EGFP cells [see Hyman et al. Proc Natl Acad Sci U S A.
2009;106(33):14132-7], an NIH-3T3-based cell line containing a stably integrated Gli-dependent enhanced green fluorescent protein reporter (Shh-EGFP) and zeocin resistance vector (pVgRXR, Invitrogen), were cultured in DMEM (Invitrogen, 11965) containing 10%
(v/v) calf serum (ATCC, 30-2030), 150 g/mL zeocin (Invitrogen, R250-01), 1 mM
sodium pyruvate (Invitrogen, 11360) and lx PenStrep (Gibco, 15140).
[0063] Wnt-LIGHT cells [see Hyman et al. Proc Natl Acad Sci U S A.
2009;106(33):14132-7], a L-cell-based cell line containing a stably integrated TCF/LCF-dependent firefly luciferase reporter (SuperTopFlash), constitutive Renilla luciferase reporter (pRLSV40, Promega), and geneticin resistance vector (pcDNA3, Invitrogen), were cultured in DMEM (Invitrogen, 11965) containing 10% (v/v) fetal bovine serum (Invitrogen, No. 26140), 400 g/mL G418 (Invitrogen, 11811) and 1X PenStrep (Gibco, 15140).
[0064] Glil null cells [see Lipinski et al. BMC Cell Biol. 2008;9:49], an immortalized mouse embryonic fibroblast cell line lacking the Glil gene, were cultured in DMEM
(Invitrogen, 11965) containing 10% (v/v) calf serum (ATCC, 30-2030), 1 mM
sodium pyruvate (Invitrogen, 11360), 1X PenStrep (Gibco, 15140) and lx MEM non-essential amino acids solution (11140, Invitrogen).
[0065] All test compounds were dissolved in DMSO at 50 mM, serially diluted by 1:3, 1:4 or 1:5 in a 96-well translucent microplate (Greiner bio-one, 650201) and stored at -20 C. These serial dilutions were added to assay media freshly with final DMSO
concentration of 0.2% (v/v).
[0066] The Sufu-KO-LIGHT cell assay provides a measure of Hh pathway activity and cytotoxicity. Sufu-KO-LIGHT cells were seeded into a 96-well plate (35,000 cells/well), cultured to confluency for 24 hrs and then incubated with test compounds in DMEM
without phenol red (Invitrogen, 26140) containing 0.5% (v/v) fetal bovine serum (Invitrogen, No. 26140), 150 g/mL zeocin (Invitrogen, R250-01), 1 mM sodium pyruvate (Invitrogen, 11360), 1X PenStrep (Gibco, 15140) for 16 hrs. Bright Glo (Promega, E2620) reagent (50 L/well) was added to the cells, and luciferase activities were determined on a microplate luminometer (Veritas). To assess cytotoxicity, after 15 hrs of incubation with test compounds, CellTiter 96 AQueous One Solution Cell Proliferation Assay (Promega, G3580) reagent (20 L/well) was added. Cell viability was determined by measuring absorption at 490 nm after 1 hr of incubation on a microplate spectrophotometer (Benchmark Plus, Bio-Rad). Biological triplicates were analyzed for each test compounds.
[0067] The Shh-LIGHT2 cell assay provides a measure of Hh pathway activity.
ShhN-conditioned medium was prepared by culturing a Shh-N-producing HEK 293 cell line [see Chen et al. Proc Natl Acad Sci USA. 2002;99(22):14071-6] in DMEM (Invitrogen, 11965) containing 10% (v/v) calf serum (ATCC, 30-2030), 1 mM sodium pyruvate (Invitrogen, 11360) and lx PenStrep (Gibco, 15140). After the cells reached 90% confluency, the medium was exchanged to DMEM (Invitrogen, 11965) containing 2% (v/v) calf serum (ATCC, 30-2030), 1 mM sodium pyruvate (Invitrogen, 11360) and 1X PenStrep (Gibco, 15140), and the resulting ShhN-conditioned medium was collected 24 hrs later and filtered through a 0.22-um membrane. Shh-LIGHT2 cells were seeded into a 96-well plate (35,000 cells/well), cultured to confluency for 32 hrs and then incubated with test compounds in DMEM (Invitrogen, 11965) containing 0.5% (v/v) calf serum (ATCC, 30-2030), 150 g/mL zeocin (Invitrogen, R250-01), 400 g/mL G418 (Invitrogen, 11811), 1 mM
sodium pyruvate (Invitrogen, 11360), 1X PenStrep (Gibco, 15140) and 10% (v/v) ShhN-conditioned medium for 30 hrs. The cells were lysed with passive lysis buffer (50 4/well), and 10 L of the obtained lysate were analyzed for firefly and Renilla luciferase activities using Dual-Luciferase Reporter Assay System (Promega, E1960) (50 4/well of each reagent) on a microplate luminometer (Veritas). Biological triplicates were analyzed for each test compounds.
[0068] The Wnt-LIGHT cell assay provides a measure of Wnt pathway activity.
Wnt3a-conditioned medium was prepared by culturing Wnt3a-expressing L cells obtained from ATCC in DMEM (Invitrogen, 11965) containing 10% (v/v) fetal bovine serum (Invitrogen, No. 26140) and 1X PenStrep (Gibco, 15140). After the cells reached 70%
confluency, the medium was exchanged to fresh medium, and the resulting Wnt3a-conditioned medium was collected 24 hrs later and filtered through a 0.22-um membrane. Wnt-LIGHT
cells were seeded into a 96-well plate (12,000 cells/well), cultured for 24 hrs and then incubated with test compounds in DMEM (Invitrogen, 11965) containing 10% (v/v) fetal bovine serum (Invitrogen, No. 26140), 50% (v/v) Wnt3a-conditioned medium, 400 g/mL G418 (Invitrogen, 11811), 1 mM sodium pyruvate (Invitrogen, 11360) and 1X PenStrep (Gibco, 15140) for 24 hrs. The cells were lysed with passive lysis buffer (50 4/well), and 10 L of the obtained lysate were analyzed for firefly and Renilla luciferase activities using Dual-Luciferase Reporter Assay System (Promega, E1960) (50 4/well of each reagent) on a microplate luminometer (Veritas). Biological triplicates were analyzed for each test compounds.
[0069] The C3H10T(1/2) cell assay provides a measure of Hh pathway-dependent osteoblast differentiation. C3H10T(1/2) cells were seeded into a 96-well plate (20,000 cells/well), cultured for 30 hrs and then incubated with test compounds in DMEM
(Invitrogen, 11965) containing 0.5% (v/v) fetal bovine serum (Invitrogen, 26140), 1X
PenStrep (Gibco, 15140), and 10% (v/v) Shh-N-conditioned medium for 44 hrs.
The cells were lysed with lysis buffer (50 L/well) containing 50 mM Tris-HC1, pH 9.5, 150 mM
NaC1, 50 mM MgC12, and 1% (v/v) Triton X-100. 10 L of the obtained lysate were analyzed for alkaline phosphatase activities using CDP-Star Substrate (Applied Biosystems, T2146) reagent (50 L/well) on a microplate luminometer (Veritas). Biological triplicates were analyzed for each test compounds.
[0070] Overexpression of G1i2 in Glil null cells provides a measure of G1i2-induced pathway activity. pcDNA-derived G1i2 expression vectors (200 ng/well) and a 1:19 mixture (total 200 ng/well) of constitutive Renilla luciferase reporter (pRLTK, Promega) and firefly luciferase reporter (8XG1iBS-FL) were mixed with FuGene HD transfection reagent (1.2 4/well, Promega, E2311) in Opti-MEM Reduced-Serum Medium with GlutaMAX
(Invitrogene, 51985) at a total volume of 25 4/well. Glil¨/¨ mouse embryonic fibroblasts were dissociated in culture medium at 800,000 cells/mL, and 1.75 mL (1,400,000 cells) were incubated with the transfection reagent-DNA mix (1.4 mL) prepared above for 3 min.
After addition of the culture medium (19.25 mL), the cells (0.4 mL/well, 25,000 cells/well) were plated into 24-well plates and cultured for 24 hrs. After the medium was exchanged to fresh medium, the cells were grown to confluence (24 hrs) and then incubated with test compounds in DMEM (Invitrogen, 11965) containing 0.5% (v/v) calf serum, lx PenStrep (Gibco, 15140) and 1 mM sodium pyruvate (Invitrogen, 11360) for 24 hrs. The cells were lysed with passive lysis buffer (200 4/well), and 10 L of the obtained lysate were analyzed for firefly and Renilla luciferase activities using Dual-Luciferase Reporter Assay System (Promega, E1960) (50 4/well of each reagent) on a microplate luminometer (Veritas). Biological triplicates were analyzed for each test compound.
[0071] Overexpression of Glil or G1i2 in NIH-3T3 cells provides a measure of Glil- or Glil/G1i2-induced pathway activity, respectively. NIH 3T3 cells were seeded into a 24-well plate (35,000 cells/well) and cultured for 30 hrs. The cells were co-transfected with pcDNA-derived Glil or G1i2 expression vectors (220 ng/well) and a 1:15 mixture (total 80 ng/well) of constitutive Renilla luciferase reporter (pRLTK, Promega) and firefly luciferase reporter (8XG1iBS-FL), using TransIT-LT1 transfection reagent (1.5 4/well, Minis Bio, MIR 2300) and Opti-MEM Reduced-Serum Medium (50 4/well, Invitrogen, 31985) according to manufacturer's protocol and cultured for 24 h. After the medium was exchanged to fresh medium, the cells were grown to confluence (24 hrs) and incubated with test compounds in DMEM (Invitrogen, 11965) containing 0.5% (v/v) calf serum, PenStrep (Gibco, 15140) and 1 mM sodium pyruvate (Invitrogen, 11360) for 24 hrs. The cells were lysed with passive lysis buffer (200 L/well), and 10 IA of the obtained lysate were analyzed for firefly and Renilla luciferase activities using Dual-Luciferase Reporter Assay System (Promega, E1960) (50 L/well of each reagent) on a microplate luminometer (Veritas). Biological triplicates were analyzed for each test compounds.
[0072] Quantification of Glil mRNA levels in NIH-3T3 cells provides a measure of endogenous Hh target gene expression. NIH 3T3 cells were seeded into 96-well plates (36,000 cells/well), cultured to confluency for 24 hrs and then incubated with test compounds in DMEM (Invitrogen, 11965) containing 0.5% (v/v) calf serum (ATCC, 2030), 1 mM sodium pyruvate (Invitrogen, 11360), 1X PenStrep (Gibco, 15140) and 10%
(v/v) ShhN-conditioned medium for 24 hrs. The cells were lysed and the lysate was used to prepare cDNA using a Cells-to-CT kit (Ambion) according to the manufacturer's protocols.
The cDNA was then quantified with Glil and GAPDH Taqman probes (Mm00494645m1 and Mm99999914g1, Applied Biosystems) on a Roche Lightcycler 480, using the 2nd derivative/maximum method to obtain Ct values. Biological triplicates were analyzed for each test compounds.
[0073] NIH 3T3 cells were also employed in a Smo trafficking assay. NIH 3T3 cells were seeded, treated with test compounds, fixed and blocked in the same way described below.
The coverslips were incubated in blocking buffer containing rabbit anti-Smo [see Rohatgi et al. Science. 2007;317(5836):372-6] and mouse monoclonal anti-N-acetylated-a-tubulin (clone 6-11B-1, 1:5000 dilution; Sigma-Aldrich, T7451) for 1 hr at room temperature. The cells were then washed three times with PBS and incubated in blocking buffer containing Alexa Fluor 488-conjugated donkey polyclonal anti-rabbit IgG antibody (1:2000 dilution;
Invitrogen, A-21206), Alexa Fluor 594-conjugated donkey polyclonal anti-mouse IgG
antibody (1:2000 dilution; Invitrogen, A-21203) for 1 hr. After washed three times with PBS, the coverlsips were mounted onto slides using Prolong Gold Antifade Reagent with DAPI (Invitrogen, P36931). The cells were imaged using a Plan Apochromat 63x/1.4-0.6 oil immersion objective on an upright Leica DM4500B compound microscope.
Ciliary Smo levels were quantified by determining total pixel intensity within a circular region manually placed at the distal end of each cilium and subtracting background fluorescence in an adjacent region of equivalent size. Ciliary Smo levels were quantified by designating ciliary regions according to N-acetylated-a-tubulin staining manually. The ciliary regions were then transferred to the corresponding images of Smo antibody staining, and the average pixel intensity was quantified by determining total pixel intensity within the region and subtracting background fluorescence in an adjacent region of equivalent size.
Total of 30 cilia from three coverslips were analyzed to determine the average ciliary Smo level for each experimental condition as a measure of Smo trafficking.
[0074] NIH 3T3 cells were also employed in an assay to quantify G1i2 trafficking. NIH-3T3 cells were seeded into 24-well plates containing poly-D-lysine-coated 12-mm glass coverslips (65,000 cells/well) and cultured for 24 hrs. The cells were then cultured in DMEM (Invitrogen, 11965) containing 0.5% (v/v) calf serum (ATCC, 30-2030), 1 mM
sodium pyruvate (Invitrogen, 11360) and 1X PenStrep (Gibco, 15140) for 20 hrs to promote primary cilia formation. The cells were next treated for 4 hrs with test compounds at concentrations 10-fold greater than their IC5Os or 0.2% (v/v) DMSO (vehicle) in the same medium described above. Each compound or vehicle treatment was conducted in biological triplicates in the presence or absence of 10% (v/v) ShhN-conditioned medium.
The cells were subsequently fixed in PBS containing 4% (v/v) paraformaldehyde for 12 min at room temperature, washed three times with PBS, permeabilized in PBS containing 0.5%
(v/v) Triton X-100 for 5 min, and washed again twice in PBS. After blocking overnight at 4 C in PBS containing 1% (w/v) bovine serum albumin (Sigma, A7030), the coverslips were incubated in blocking buffer containing goat polyclonal anti-G1i2 antibody (1:150 dilution;
R & D Systems, AF3635), and mouse monoclonal anti-N-acetylated-a-tubulin (clone 6-11B-1, 1:5000 dilution; Sigma-Aldrich, T7451) for 1 hr at room temperature.
The cells were then washed three times with PBS and incubated in blocking buffer containing DyLight 488-conjugated donkey polyclonal anti-goat IgG antibody (3 ug/mL; Jackson ImmunoResearch, 705-485-147), DyLight 594-conjugated donkey polyclonal anti-mouse IgG antibody (3 ug/mL; Jackson ImmunoResearch, 715-515-151) for 1 hr. After three PBS
washes, the coverslips were mounted onto slides using Prolong Gold Antifade Reagent with DAPI (Invitrogen, P36931). The cells were imaged using a Plan Apochromat 63x/1.4-0.6 oil immersion objective on an upright Leica DM4500B compound microscope.
Ciliary G1i2 levels were quantified by determining total pixel intensity within a circular region manually placed at the distal end of each cilium and subtracting background fluorescence in an adjacent region of equivalent size. A total of 30 cilia from three coverslips were analyzed to determine the average ciliary G1i2 level for each experimental condition as a measure of G1i2 trafficking.
[0075] A quantitative assessment of Hh ligand-dependent G1i3 processing was provided by the following assay: Shh-EGFP cells were seeded into 12-well plates at a density of 160,000 cells/well and cultured for 24 hrs in DMEM (Invitrogen, 11965) containing 10%
(v/v) calf serum (ATCC, 30-2030), 150 g/mL zeocin (Invitrogen, R250-01), 1 mM
sodium pyruvate (Invitrogen, 11360) and 1X PenStrep (Gibco, 15140). The cells were next treated for 16 hrs with test compounds at concentrations 10-fold greater than their IC5Os or 0.2%
(v/v) DMSO (vehicle) in DMEM (Invitrogen, 11965) containing 0.5% (v/v) calf serum (ATCC, 30-2030) and antibiotics described above. Each compound or vehicle treatment was conducted in the presence or absence of 10% (v/v) Shh-N-conditioned medium. The cells were then lysed by incubation with SDS-PAGE loading buffer composed of 50 mM
Tris-HC1, pH 6.8, 2% (w/v) SDS, 8% (v/v) glycerol, 100 mM DTT, 0.1 mg/mL
bromophenol blue, EDTA-free protease inhibitor cocktail (Roche) and Phosstop (Roche) for min in a cold room. The lysate were heated to 100 C for 7 min, loaded onto 3-8%
Criterion XT Tris-Acetate polyacrylamide gels (Bio-Rad, 345-0129), electrophoresed in XT
Tricine buffer (Bio-Rad, 161-0790), and transferred onto PVDF membranes (EMD
Millipore, IPVH304F0). The membranes were dehydrated with methanol and probed overnight at 4 C with goat polyclonal anti-G1i3 antibody (1 gg/mL; R & D
Systems, AF3690) in PBS containing 4% (w/v) non-fat dry milk and 0.01% (v/v) Tween-20 (immunoblot blocking buffer). The blots were then washed four times in PBS and incubated with horseradish peroxidase-conjugated bovine polyclonal anti-goat IgG
antibody (0.16 gg/mL; Jackson ImmunoResearch, 805-035-180) in immunoblot blocking buffer for 1 hr at room temperature. The membranes were next washed four times in PBS and visualized using SuperSignal West Dura Extended Duration substrate (Thermo Scientific, 34076) and a ChemiDoc XRS imaging system (Bio-Rad). Band intensities of G1i3FL and G1i3R
were quantified using ImageJ64 software (NIH), and three independent experiments were used to determine the average G1i3FL/G1i3R ratio for each compound.
[0076]
Cell line Reporter ICso (11M) Sufu-KO-LIGHT firefly luciferase 1.42 0.96 Sufu-KO-LIGHT cytotoxicity No toxicity Wnt-LIGHT w/ Wnt3a firefly luciferase No inhibition Shh-LIGHT2 w/ ShhN firefly luciferase 6.6 C3H10T(1/2) w/ ShhN alkaline phosphatase 5.2 3.7 NIH-3T3 w/ ShhN Glil mRNA 5 Glii-overexpressing NIH-3T3 firefly luciferase 0.16 G/i2-overexpressing NIH-3T3 firefly luciferase 0.77 0.88 G/i2-overexpressing Gli1-1- firefly luciferase No inhibition
[0077] The foregoing studies confirmed that A-1 inhibits Hh pathway activity in Sufu-KO-LIGHT cells, as well as NIH-3T3 cells stably transfected with the Gli-dependent firefly luciferase reporter (Shh-LIGHT2 cells) and stimulated with medium containing the Shh N-terminal domain (ShhN). The compound blocked the ability of ShhN-conditioned medium to differentiate C3H10T1/2 cells into alkaline-phosphatase-expressing osteoblasts and inhibited the ShhN-dependent expression of endogenous target genes (G111).
Studies also confirmed that A-1 was not generally cytotoxic and that it did not inhibit Wnt signaling compound.
[0078] The ability of A-1 to inhibit Hh reporter in Sufu-KO-LIGHT cells indicates that it acts at the level of the Gli transcription factors within the pathway.
Consistent with this downstream site of action, the compound did not affect Smo localization in Hh-responsive cells. ShhN stimulation causes Smo to accumulate within the primary cilium, and the known Smo inhibitor cyclopamine also alters Smo trafficking. A-1, however, did not affect Smo localization. Similarly, ShhN treatment causes G1i2 to accrue at the ciliary distal tip in a cyclopamine-sensitive manner, but A-1 did not inhibit this process. These results suggest that A-1 does not prevent Gli transcription factor activation. While ShhN
stimulation caused the G1i3FL/G1i3R ratio to increase and cyclopamine abrogated that effect, A-1 did not perturb G1i3 processing. Nor did the compound prevent G1i3FL
phosphorylation.
[0079] Compound A-1 potently blocked exogenous Glil activity but only partially inhibited Hh reporter expression induced by exogenous G1i2. Since Glil is a Hh target gene that will be expressed in response to G1i2 overexpression, it is reasonable to assume that this partial effect could reflect action of A-1 on Glil but not G1i2. Consistent with this model, A-1 was not able to inhibit G1i2-induced Hh reporter expression in cells lacking Glil. Thus, the evidence indicates that A-1 is representative of a class of specific inhibitors of Glil.
Representative results of studies in vitro on other species within the genus I
are outlined in Tables 1 to 3.

N
R1 R2a _________________________________ R3a Comp. ICso (LIM) Br A-1 1.42 0.96 Et0 4104 Br A-2 7.9 40 Br A-3 0.34 OMe 0 qt.H Br A-4 5.0 c.-0 1104 Ri R2a R3a _______________________ Comp. 'Cs() (jM) = 1_ H
104 Br A-5 12.3 F3c ci = v H . Br A-6 11.7 CI A-7 3.0 Et0 4. I- H

Et0 H
A-8 0.19 0.14 4. I-Et0 4. I- H . A-9 1.7 Et0 . H 0 OMe A-10 2.5 CI A-11 2.5 meo '.' I- H
0CF2H A-12 13.9 meo 4, I- H
*
H CI A-13 17.5 Ri R2a R3a Comp. 'Cs() (jM) Br lik 1- H . OMe A-15 3.3 = 1_ H * OEt A-16 7.4 OCF2H A-17 2.9 Et 4.' 1- H
fieOEt A-18 0.08 OMe . V H *I . A-19 0.03 OMe '?

R3' R \........... X-N
VN*------Ri R2a R3a Comp. ICso (11M) %_ Me0 H C-1 1.6 Et0 = %
lik . 1¨ Me0 H C-2 3.1 OMe 1111 Ri R2a R3a Comp. ICso (11-11\4) 4. 1... Me0 H C-3 4.2 IP
L. Me0 H C-4 11.5 CI =
lik En Me C-5 1.2 Eta . z lik %_ CI
Me0 H C-6 11.7 = z lik 4. µ_ HF2C0 H C-7 6.3 Me0 IP
Me0 = µ- 111, H C-8 4.1 Ci 411i 1¨ CI
IIP H C-9 27.0 Structure Comp. ICso (11M) Et0 D-1 0.96 N+ N
Me0 410 OMe D-2 3.1 N+ N
)\¨N
SN) Me0 D-3 1.1 N+ N
)\¨N
SN) Me0 Br D-4 2.6 N+ N
)\¨N
SN) Me0 CI D-5 2.4 N+ N
)\¨N
S\
Me0 110 * OMe D-6 14.1 NJ+ N
tN
Oi OEt E-1 0.14 as *
Br-Me0
[0080] The foregoing results in vitro predict utility against Hh pathway-dependent tumors. Representative compounds A-8 and E-1 have been tested in vivo.
[0081] Murine basal cell carcinomas (BCCs) were derived from Ptchl+/¨;p53¨/¨
mice subjected to UV radiation [Aszterbaum et al., Nat. Med. (1999) 5:1285-91]. A
line resistant to the Smo antagonist SANT-1 was obtained by culturing the cells in media containing increasing concentrations of the inhibitor until the majority of cells were killed and resistant clones emerged.
[0082] Murine BCC allograft model: Ptchl+/¨;K14:Cre-ER2;p53fl/fl mice were injected with tamoxifen (to knock out p53) at six weeks of age, and at 8 weeks of age they were given one dose of 4 Gy of ionizing radiation. After BCCs formed and grew to 5-7 mm in diameter (typically between 6-8 months of age), they were harvested and dissociated to prepare a cell suspension. A mixture of the cell suspension and Matrigel was then injected subcutaneously into NOD/SCID mice purchased from Jackson Laboratories. Once the allografted tumors became visible, they were monitored with digital calipers and the volume was estimated by the formula (d'2) *D/2, where d is the smallest diameter of the tumor and D is the largest diameter. The compound was applied topically to the tumor as a DMSO
solution (100 iut per application).
[0083] Murine medulloblastoma allograft model: Ptchl+/¨;p53+/¨;Mathl:GFP mice were mated to generate pups with the appropriate genotype of Ptchl+/--;p53¨/¨

;Mathl :GFP. These mice will get medulloblastoma tumors when they are 6 to 10 weeks of age. The primary medulloblastoma tumors were harvested, dissected, dissociated by papain treatment and tissue trituration, and injected into the flanks of nude mice as a Matrigel mixture (50 iut of 2 x 107 tumor cells mixed with 50 iut of Matrigel).
Secondary and tertiary tumors can subsequently be derived from these allografts as necessary. The length, width, and depth of each allograft were monitored, and compound testing began when the tumors reached a volume of 150 mm3 (approximately one week after tumors were first visible). The compounds were dissolved in a minimum volume of DMSO, and the DMSO
solution was mixed with sterile corn oil so that the DMSO constituted less than 10% of the total volume. The drug was administered by intraperitoneal injection (50 L), with each daily dose divided into two portions and administered at 12-hour intervals.
The Smoothened antagonist vismodegib was administered at 50 mg/kg/day as a positive control.
[0084] Compound A-8 reduced BCC proliferation in dose-dependent fashion to 20%
of control at 750 nM. A 0.01% solution reduced tumor volume by half in the murine BCC
allograft model compared to control at day 14. Compound A-8 was able to block the proliferation of both the original BCC cell line and the SANT-1-resistant line. This indicates that these compounds could be used against tumors that have acquired resistance to Smo inhibitors. Compound E-1 at 2 mg/kg/day in the murine medulloblastoma allograft model reduced tumor volume by half compared to control at day 5.

Claims (23)

1. Use of a compound of formula I
for inhibiting the growth of a solid tumor wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2 is chosen from H, halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, optionally substituted aryl and optionally substituted heteroaryl;
R3 is chosen from H, alkyl, optionally substituted aryl and optionally substituted heteroaryl;
A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (C1-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (C1-C4)alkyl residues;
and X is any counterion;
with the proviso that one of R2 and R3 must be optionally substituted aryl or optionally substituted heteroaryl.
2. Use of a compound of formula I

for inhibiting Hedgehog pathway activation wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2 is chosen from H, halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, optionally substituted aryl and optionally substituted heteroaryl;
R3 is chosen from H, alkyl, optionally substituted aryl and optionally substituted heteroaryl;
A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (C1-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (C1-C4)alkyl residues;
and X is any counterion;
with the proviso that one of R2 and R3 must be optionally substituted aryl or optionally substituted heteroaryl.
3. Use of a compound of formula I

for probing Hedgehog function in vitro wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2 is chosen from H, halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, optionally substituted aryl and optionally substituted heteroaryl;
R3 is chosen from H, alkyl, optionally substituted aryl and optionally substituted heteroaryl;
A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (C1-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (C1-C4)alkyl residues;
and X is any counterion;
with the proviso that one of R2 and R3 must be optionally substituted aryl or optionally substituted heteroaryl.
4. Use according to any of claims 1 to 3 wherein ring A is chosen from pyrrolidine, piperidine, azepine, thiazolidine, oxazolidine, imidazolidine, thiazine, oxazine, piperazine, oxazepine, thiazepine, diazepine and tetrahydroquinoline.
5. Use according to claim 4 wherein ring A is chosen from pyrrolidine, piperidine, azepine, thiazolidine, thiazine, morpholine and tetrahydroquinoline.
6. Use according to any of claims 1 to 3 wherein R1 is phenyl or optionally substituted phenyl and one of R2 and R3 is phenyl or optionally substituted phenyl.
7. Use according to claim 6 wherein R1 is phenyl or phenyl substituted with a substituent chosen from halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, (C1-C4)alkoxy, halo(C1-C4)alkoxy, (C1-C2)alkylenedioxy and phenyl.
8. Use according to claim 7 wherein R2 is H or methyl and R3 is phenyl or phenyl substituted with a substituent chosen from halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, (C1-C4)alkoxy, halo(C1-C4)alkoxy, (C1-C2)alkylenedioxy, and phenyl.
9. Use according to claim 7 wherein R3 is H or methyl and R2 is phenyl or phenyl substituted with a substituent chosen from halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, (C1-C4)alkoxy, halo(C1-C4)alkoxy, (C1-C2)alkylenedioxy, and phenyl.
10. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of formula II:
wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2a is chosen from H and (C1-C4)alkyl;
R3a is chosen from optionally substituted aryl and optionally substituted heteroaryl;
A1 is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (C1-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (C1-C4)alkyl residues;
and X is any counterion.
11. A pharmaceutical composition according to claim 10 wherein ring A is chosen from piperidine, azepine, thiazine, oxazine, piperazine, oxazepine, thiazepine, diazepine and tetrahydroquinoline.
12. A pharmaceutical composition according to claim 10 wherein II has the formula IIa:
13. A pharmaceutical composition according to claim 10 wherein II has the formula IIb:
14. A pharmaceutical composition according to claim 10 wherein II has the formula IIc:

15. A
pharmaceutical composition according to any of claims 10 to 14 wherein R1 is phenyl or optionally substituted phenyl and R3a is phenyl or optionally substituted phenyl.
16. A pharmaceutical composition according to claim 15 wherein R1 is phenyl or phenyl substituted with a substituent chosen from halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, (C1-C4)alkoxy, halo(C1-C4)alkoxy, (C1-C2)alkylenedioxy and phenyl.
17.A pharmaceutical composition according to claim 16 wherein R2a is H or methyl and R3a is phenyl or phenyl substituted with a substituent chosen from halogen, (C1-C4)alkyl, halo (C1-C4)alkyl, (C1-C4)alkoxy, halo (C1-C4)alkoxy, (C1-C2)alkylenedioxy, and phenyl.
18. A compound of formula wherein A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (C1-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (C1-C4)alkyl residues;
R6 is one or two substituents chosen independently from the group consisting of halogen, halo (C1-C6)alkyl, (C1-C6)alkyl, (C1-C6)acyl, (C1-C6)alkoxy(C1-C6)alkyl, hydroxy(C1-C6)alkyl, phenyl, benzenesulfonyl, hydroxy, (C1-C6)alkoxy, halo(C1-C6)alkoxy, oxaalkyl, carboxy, (C1-C6)alkoxycarbonyl , (C1-C6)alkoxycarbonylamino, carboxamido, cyano, acetoxy, nitro, amino, (C1-C6)alkylamino, (C1-C6)dialkylamino, mercapto, (C1-C6)alkylthio, (C1-C6)alkylsulfoxide, (C1-C6)alkylsulfonyl, benzyl, heterocyclyl, phenoxy, heteroaryloxy and benzyloxy, and X is any counterion;
with the proviso that when A is azepine and R6 is bromine or methoxy, R6 is not in the para position.
19. A compound according to claim 18 of formula:
20. A compound according to claim 19 wherein R6 is chosen from ethoxy and phenyl.
21. A method of inhibiting Hedgehog pathway activation comprising bringing a cell that is capable of Hedgehog expression into contact with a compound of formula I

wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2 is chosen from H, halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, optionally substituted aryl and optionally substituted heteroaryl;
R3 is chosen from H, alkyl, optionally substituted aryl and optionally substituted heteroaryl;
A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (C1-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (C1-C4)alkyl residues;
and X is any counterion;
with the proviso that one of R2 and R3 must be optionally substituted aryl or optionally substituted heteroaryl.
22. A method of inhibiting the growth of a solid tumor comprising bringing said solid tumor into contact with a compound of formula wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2 is chosen from H, halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, optionally substituted aryl and optionally substituted heteroaryl;
R3 is chosen from H, alkyl, optionally substituted aryl and optionally substituted heteroaryl;
A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (C1-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (C1-C4)alkyl residues;
and X is any counterion;
with the proviso that one of R2 and R3 must be optionally substituted aryl or optionally substituted heteroaryl.
23. A method of probing Hedgehog function in vitro comprising bringing a cell that is capable of Hedgehog expression into contact with a compound of formula wherein:
R1 is chosen from optionally substituted aryl and optionally substituted heteroaryl;
R2 is chosen from H, halogen, (C1-C4)alkyl, halo(C1-C4)alkyl, optionally substituted aryl and optionally substituted heteroaryl;
R3 is chosen from H, alkyl, optionally substituted aryl and optionally substituted heteroaryl;
A is (a) a fused, saturated ring of 5 to 7 members that may contain other heteroatoms in addition the nitrogen at the point of fusion, said saturated ring optionally substituted with one or two (C1-C4)alkyl residues or (b) a fused bicycle, at least one ring of said fused bicycle being non-aromatic and said bicycle optionally substituted with one or two (C1-C4)alkyl residues;
and X is any counterion;
with the proviso that one of R2 and R3 must be optionally substituted aryl or optionally substituted heteroaryl.
CA2877473A 2012-06-22 2013-06-19 Imidazo bicyclic imminium compounds as antitumor agents Abandoned CA2877473A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201261662977P 2012-06-22 2012-06-22
US61/662,977 2012-06-22
PCT/US2013/046560 WO2013192301A1 (en) 2012-06-22 2013-06-19 Imidazo bicyclic imminium compounds as antitumor agents

Publications (1)

Publication Number Publication Date
CA2877473A1 true CA2877473A1 (en) 2013-12-27

Family

ID=49769330

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2877473A Abandoned CA2877473A1 (en) 2012-06-22 2013-06-19 Imidazo bicyclic imminium compounds as antitumor agents

Country Status (5)

Country Link
US (1) US9611276B2 (en)
EP (1) EP2864332B1 (en)
AU (1) AU2013277199A1 (en)
CA (1) CA2877473A1 (en)
WO (1) WO2013192301A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4167995A1 (en) * 2020-06-18 2023-04-26 The Board of Trustees of the Leland Stanford Junior University Isoform-specific aldehyde dehydrogenase inhibitors

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2942000A (en) 1999-03-11 2000-09-28 Banyu Pharmaceutical Co., Ltd. Novel piperidine derivatives
GB0517966D0 (en) 2005-09-02 2005-10-12 Novartis Ag Organic compounds
WO2008064136A2 (en) * 2006-11-17 2008-05-29 Acadia Pharmaceuticals Inc. Compounds with activity at retinoic acid receptors
US8486936B2 (en) 2009-05-05 2013-07-16 AllaChem, LLC Antagonist of smoothened

Also Published As

Publication number Publication date
EP2864332A1 (en) 2015-04-29
WO2013192301A1 (en) 2013-12-27
EP2864332B1 (en) 2016-12-28
EP2864332A4 (en) 2015-05-06
US20150191489A1 (en) 2015-07-09
US9611276B2 (en) 2017-04-04
AU2013277199A1 (en) 2015-01-29

Similar Documents

Publication Publication Date Title
EP3440067B1 (en) Tetrahydroisoquinoline estrogen receptor modulators and uses thereof
AU2015276537B2 (en) Novel compounds as dual inhibitors of histone methyltransferases and dna methyltransferases
ES2798424T3 (en) Triazolopyridine Compounds and Uses of These
JP6640222B2 (en) Heterocyclic derivatives and uses thereof
US20130281396A1 (en) Treatment of diseases by epigenetic regulation
US9573899B2 (en) USP7 inhibitor compounds and methods of use
KR20080007443A (en) Thienopyridine derivative, or quinoline derivative, or quinazoline derivative, having c-met autophosphorylation inhibiting potency
KR20090119768A (en) Treatment of melanoma
JP2018138609A (en) Naphthyridinedione derivatives
JP2011526912A (en) Naphthyridinone as a protein kinase inhibitor
CN113164776A (en) Tyrosine kinase inhibitor compositions, methods of making and methods of using the same
WO2021257697A1 (en) Compounds and methods for blocking apoptosis and inducing autophagy
US9611276B2 (en) Imidazo bicyclic iminium compounds as antitumor agents
US9145376B2 (en) Quinazolinone inhibitors of dynein
JP2017516829A (en) Pyridopyrimidinedione derivatives
US11773096B2 (en) Small-molecule PI5P4K alpha/beta inhibitors and methods of treatment using same
RU2480469C1 (en) Substituted 1,3-diethyl-8-vinyl-7-methyl-3,7-dihydropurine-2,6-diones - antagonists of adenosine a2a receptor and use thereof
NZ730448A (en) TETRAHYDRO-PYRIDO[3,4-b]INDOLE ESTROGEN RECEPTOR MODULATORS AND USES THEREOF
NZ730448B2 (en) TETRAHYDRO-PYRIDO[3,4-b]INDOLE ESTROGEN RECEPTOR MODULATORS AND USES THEREOF
CA3006413A1 (en) Benzo[b][1,6]naphthyridine derivatives as cpap-tubulin inhibitors
JP2011102252A (en) IMIDAZO[1,2-a]BENZIMIDAZOLE COMPOUND HAVING ERYTHROPOIETIN PRODUCTION-PROMOTING EFFECT
JP2011105659A (en) INDOLO[2,3-b]QUINOXALINE COMPOUND HAVING ERYTHROPOIETIN PRODUCTION-PROMOTING EFFECT

Legal Events

Date Code Title Description
FZDE Dead

Effective date: 20180619